Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Arnau Montraveta is active.

Publication


Featured researches published by Arnau Montraveta.


Leukemia | 2014

Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma

Alexandra Moros; Vanina Rodriguez; Ifigènia Saborit-Villarroya; Arnau Montraveta; Patricia Balsas; P Sandy; Antoni Martínez; Adrian Wiestner; Emmanuel Normant; Elias Campo; Patricia Pérez-Galán; Dolors Colomer; Gaël Roué

Bortezomib therapy has shown promising clinical activity in mantle cell lymphoma (MCL), but the development of resistance to proteasome inhibition may limit its efficacy. To unravel the factors involved in the acquisition of bortezomib resistance in vivo, immunodeficient mice were engrafted with a set of MCL cell lines with different levels of sensitivity to the drug, followed by gene expression profiling of the tumors and functional validation of the identified gene signatures. We observed an increased tumorigenicity of bortezomib-resistant MCL cells in vivo, which was associated with plasmacytic differentiation features, like interferon regulatory factor 4 (IRF4) and Blimp-1 upregulation. Lenalidomide was particularly active in this subgroup of tumors, targeting IRF4 expression and plasmacytic differentiation program, thus overcoming bortezomib resistance. Moreover, repression of the IRF4 target gene MYC in bortezomib-resistant cells by gene knockdown or treatment with CPI203, a BET (bromodomain and extra terminal) bromodomain inhibitor, synergistically induced cell death when combined with lenalidomide. In mice, addition of CPI203 to lenalidomide therapy further decreased tumor burden, involving simultaneous MYC and IRF4 downregulation and apoptosis induction. Together, these results suggest that exacerbated IRF4/MYC signaling is associated to bortezomib resistance in MCL in vivo and warrant clinical evaluation of lenalidomide plus BET inhibitor combination in MCL cases refractory to proteasome inhibition.


Leukemia | 2015

The γ-secretase inhibitor PF-03084014 combined with fludarabine antagonizes migration, invasion and angiogenesis in NOTCH1-mutated CLL cells

Mónica López-Guerra; Sílvia Xargay-Torrent; Laia Rosich; Arnau Montraveta; J Roldán; A Matas-Céspedes; Neus Villamor; M. Aymerich; Carlos López-Otín; Patricia Pérez-Galán; Gaël Roué; Elias Campo; Dolors Colomer

Targeting Notch signaling has emerged as a promising therapeutic strategy for chronic lymphocytic leukemia (CLL), especially for the poor prognostic subgroup of NOTCH1-mutated patients. Here, we report that the γ-secretase inhibitor PF-03084014 inhibits the constitutive Notch activation and induces selective apoptosis in CLL cells carrying NOTCH1 mutations. Combination of PF-03084014 with fludarabine has a synergistic antileukemic effect in primary NOTCH1-mutated CLL cells, even in the presence of the protective stroma. At transcriptional level, PF-03084014 plus fludarabine treatment induces the upregulation of the proapoptotic gene HRK and the downmodulation of MMP9, IL32 and RAC2 genes that are related to invasion and chemotaxis. PF-03084014 also overcomes fludarabine-mediated activation of nuclear factor-κB signaling. Moreover, this combination impairs angiogenesis and CXCL12-induced responses in NOTCH1-mutated CLL cells, in particular those related to tumoral migration and invasion. Importantly, all these collaborative effects are specific for NOTCH1 mutation and do not occur in unmutated cases. In conclusion, we provide evidence that Notch is a therapeutic target in CLL cases with NOTCH1-activating mutations, supporting the use of Notch pathway inhibitors in combination with chemotherapy as a promising approach for the treatment of these high-risk CLL patients.


Haematologica | 2013

The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/FoxO3a/Bim axis in chronic lymphocytic leukemia cells

Laia Rosich; Ifigènia Saborit-Villarroya; Mónica López-Guerra; Sílvia Xargay-Torrent; Arnau Montraveta; Marta Aymerich; Neus Villamor; Elias Campo; Patricia Pérez-Galán; Gaël Roué; Dolors Colomer

Phosphatidylinositol-3-kinase pathway is constitutively activated in chronic lymphocytic leukemia mainly due to microenvironment signals, including stromal cell interaction and CXCR4 and B-cell receptor activation. Because of the importance of phosphatidylinositol-3-kinase signaling in chronic lymphocytic leukemia, we investigated the activity of the NVP-BKM120, an orally available pan class I phosphatidylinositol-3-kinase inhibitor. Sensitivity to NVP-BKM120 was analyzed in chronic lymphocytic leukemia primary samples in the context of B-cell receptor and microenvironment stimulation. NVP-BKM120 promoted mitochondrial apoptosis in most primary cells independently of common prognostic markers. NVP-BKM120 activity induced the blockage of phosphatidylinositol-3-kinase signaling, decreased Akt and FoxO3a phosphorylation leading to concomitant Mcl-1 downregulation and Bim induction. Accordingly, selective knockdown of BIM rescued cells from NVP-BKM120-induced apoptosis, while the kinase inhibitor synergistically enhanced the apoptosis induced by the BH3-mimetic ABT-263. We also found NVP-BKM120 to inhibit B-cell receptor- and stroma-dependent Akt pathway activation, thus sensitizing chronic lymphocytic leukemia cells to bendamustine and fludarabine. Furthermore, NVP-BKM120 down-regulated secretion of chemokines after B-cell receptor stimulation and inhibited cell chemotaxis and actin polymerization upon CXCR4 triggering by CXCL12. Our findings establish that NVP-BKM120 effectively inhibits the phosphatidylinositol-3-kinase signaling pathway and disturbs the protective effect of the tumor microenvironment with the subsequent apoptosis induction through the Akt/FoxO3a/Bim axis. We provide here a strong rationale for undertaking clinical trials of NVP-BKM120 in chronic lymphocytic leukemia patients alone or in combination therapies.


Clinical Cancer Research | 2017

The Bruton's tyrosine kinase (BTK) inhibitor acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia

Sarah E.M. Herman; Arnau Montraveta; Carsten U. Niemann; Helena Mora-Jensen; Michael Gulrajani; Fanny Krantz; Rose Mantel; Lisa L. Smith; Fabienne McClanahan; Bonnie K. Harrington; Dolors Colomer; Todd Covey; John C. Byrd; Raquel Izumi; Allard Kaptein; Roger Ulrich; Amy J. Johnson; Brian Lannutti; Adrian Wiestner; Jennifer A. Woyach

Purpose: Acalabrutinib (ACP-196) is a novel, potent, and highly selective Bruton tyrosine kinase (BTK) inhibitor, which binds covalently to Cys481 in the ATP-binding pocket of BTK. We sought to evaluate the antitumor effects of acalabrutinib treatment in two established mouse models of chronic lymphocytic leukemia (CLL). Experimental Design: Two distinct mouse models were used, the TCL1 adoptive transfer model where leukemic cells from Eμ-TCL1 transgenic mice are transplanted into C57BL/6 mice, and the human NSG primary CLL xenograft model. Mice received either vehicle or acalabrutinib formulated into the drinking water. Results: Utilizing biochemical assays, we demonstrate that acalabrutinib is a highly selective BTK inhibitor as compared with ibrutinib. In the human CLL NSG xenograft model, treatment with acalabrutinib demonstrated on-target effects, including decreased phosphorylation of PLCγ2, ERK, and significant inhibition of CLL cell proliferation. Furthermore, tumor burden in the spleen of the mice treated with acalabrutinib was significantly decreased compared with vehicle-treated mice. Similarly, in the TCL1 adoptive transfer model, decreased phosphorylation of BTK, PLCγ2, and S6 was observed. Most notably, treatment with acalabrutinib resulted in a significant increase in survival compared with mice receiving vehicle. Conclusions: Treatment with acalabrutinib potently inhibits BTK in vivo, leading to on-target decreases in the activation of key signaling molecules (including BTK, PLCγ2, S6, and ERK). In two complementary mouse models of CLL, acalabrutinib significantly reduced tumor burden and increased survival compared with vehicle treatment. Overall, acalabrutinib showed increased BTK selectivity compared with ibrutinib while demonstrating significant antitumor efficacy in vivo on par with ibrutinib. Clin Cancer Res; 23(11); 2831–41. ©2016 AACR.


Clinical Cancer Research | 2013

Sorafenib inhibits cell migration and stroma-mediated bortezomib resistance by interfering B-cell receptor signaling and protein translation in mantle cell lymphoma

Sílvia Xargay-Torrent; Mónica López-Guerra; Arnau Montraveta; Ifigènia Saborit-Villarroya; Laia Rosich; Alba Navarro; Patricia Pérez-Galán; Gaël Roué; Elias Campo; Dolors Colomer

Purpose: We evaluated the antitumoral properties of the multikinase inhibitor sorafenib in mantle cell lymphoma (MCL), an aggressive B lymphoma for which current therapies have shown limited efficacy. Experimental Design: Sensitivity to sorafenib was analyzed in MCL cell lines and primary samples in the context of BCR and microenvironment simulation. Sorafenib signaling was characterized by quantitative PCR, Western blotting, immunofluorescence, and protein immunoprecipitation. Migration analysis included flow cytometric counting, actin polymerization assays, and siRNA-mediated knockdown of focal adhesion kinase (FAK). In vivo antitumor effect of sorafenib and bortezomib was analyzed in an MCL xenograft mouse model. Results: Sorafenib rapidly dephosphorylates the BCR-associated kinases, Syk and Lyn, as well as FAK, an Src target involved in focal adhesion. In this line, sorafenib displays strong synergy with the Syk inhibitor, R406. Sorafenib also blocks Mcl-1 and cyclin D1 translation, which promotes an imbalance between pro- and antiapoptotic proteins and facilitates Bax release from cyclin D1, leading to the induction of mitochondrial apoptosis and caspase-dependent and -independent mechanisms. Moreover, sorafenib inhibits MCL cell migration and CXCL12-induced actin polymerization. FAK knockdown partially prevents this inhibitory effect, indicating that FAK is a relevant target of sorafenib. Furthermore, sorafenib enhances the antitumoral activity of bortezomib in an MCL xenograft mouse model as well as overcomes stroma-mediated bortezomib resistance in MCL cells. Conclusion: We show for the first time that sorafenib interferes with BCR signaling, protein translation and modulates the microenvironment prosurvival signals in MCL, suggesting that sorafenib, alone or in combination with bortezomib, may represent a promising approach to treat patients with MCL. Clin Cancer Res; 19(3); 586–97. ©2012 AACR.


Pharmacogenomics Journal | 2015

Human organic cation transporter 1 (hOCT1) as a mediator of bendamustine uptake and cytotoxicity in chronic lymphocytic leukemia (CLL) cells.

Cristina Arimany-Nardi; Arnau Montraveta; Eriong Lee-Vergés; Xose S. Puente; H Koepsell; Elias Campo; Dolors Colomer; Marçal Pastor-Anglada

Bendamustine is used in the treatment of chronic lymphocytic leukemia (CLL). Routes for bendamustine entry into target cells are unknown. This study aimed at identifying transporter proteins implicated in bendamustine uptake. Our results showed that hOCT1 is a bendamustine transporter, as bendamustine could cis-inhibit the uptake of a canonical hOCT1 substrate, with a Ki in the micromolar range, consistent with the EC50 values of the cytotoxicity triggered by this drug in HEK293 cells expressing hOCT1. hOCT1 polymorphic variants determining impaired bendamustine-transporter interaction, consistently reduced bendamustine cytotoxicity in HEK293 cells stably expressing them. Exome genotyping of the SLC22A1 gene, encoding hOCT1, was undertaken in a cohort of 241 CLL patients. Ex vivo cytotoxicity to bendamustine was measured in a subset of cases and shown to correlate with SLC22A1 polymorphic variants. In conclusion, hOCT1 is a suitable bendamustine transporter, thereby contributing to its cytotoxic effect depending upon the hOCT1 genetic variants expressed.


Cancer Research | 2014

Abstract 2624: The novel Bruton's tyrosine kinase inhibitor ACP-196 shows in vivo efficacy against human chronic lymphocytic leukemia cells xenografted to the NSG mouse model

Carsten U. Niemann; Arnau Montraveta; Sarah E. M. Herman; Tim Ingallinera; Tjeerd Barf; Dolors Colomer; Adrian Wiestner

Background: Targeting Bruton9s tyrosine kinase (BTK), an essential kinase in the B cell receptor (BCR) pathway in patients with chronic lymphocytic leukemia (CLL) has proven very effective. For the first generation BTK inhibitor ibrutinib, clinical response rates >70% and 75% progression free survival >2 years have been reported for previously treated patients (Byrd, NEJM, 2013). ACP-196 is a novel, second generation, irreversible BTK inhibitor that may show advantages in terms of binding specificity and drug-drug interactions compared to the first generation BTK inhibitor. Survival and proliferation of CLL cells is highly dependent on microenvironment interaction, which must be taken into consideration when testing new drugs for CLL. We have previously shown that human CLL cells engrafted in the spleen of NSG mice have comparable tumor biology to human lymph node resident CLL cells (Herman, Leukemia, 2013). We here demonstrate the in vivo effects of ACP-196 monotherapy against CLL cells in the NSG xenograft model. Methods: Peripheral blood mononuclear cells from previously untreated CLL patients were injected intravenously into NSG mice. Mice received ACP-196 through the drinking water. The effect of ACP-196 on xenografted CLL cells from peripheral blood and spleen was assessed by flow cytometry. Results: At all dose levels tested, ACP-196 significantly inhibited proliferation of human CLL cells in the spleens of NSG mice, as measured by Ki67 expression (P=0.002). The mean Ki67 decrease was 58%, 70% and 73%, respectively for the low, intermediate, and high dose level compared with vehicle. Tumor burden in spleens from mice treated with the high dose of ACP-196 decreased compared with the vehicle treated group (P=0.007). As seen with other BCR inhibitors, ACP-196 transiently increased CLL cell counts in the peripheral blood in a dose dependent manner (p=0.01). ACP-196 inhibited BCR signaling in vivo, as demonstrated by reduced phosphorylation of PLCγ2. Conclusions: Results presented here for ACP-196 are in accordance with the expected profile of an irreversible BTK inhibitor. The NSG CLL xenograft model will be used for comparative testing of different BTK inhibitors and exploration of combination therapies. Citation Format: Carsten U. Niemann, Arnau Montraveta, Sarah E. M. Herman, Tim Ingallinera, Tjeerd Barf, Dolors Colomer, Adrian Wiestner. The novel Bruton9s tyrosine kinase inhibitor ACP-196 shows in vivo efficacy against human chronic lymphocytic leukemia cells xenografted to the NSG mouse model. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2624. doi:10.1158/1538-7445.AM2014-2624


Oncotarget | 2016

CD69 expression potentially predicts response to bendamustine and its modulation by ibrutinib or idelalisib enhances cytotoxic effect in chronic lymphocytic leukemia

Arnau Montraveta; Eriong Lee-Vergés; Jocabed Roldán; Laura Jiménez; Sandra Cabezas; Guillem Clot; Magda Pinyol; Sílvia Xargay-Torrent; Laia Rosich; Cristina Arimany-Nardi; Marta Aymerich; Neus Villamor; Armando López-Guillermo; Patricia Pérez-Galán; Gaël Roué; Marçal Pastor-Anglada; Elias Campo; Mónica López-Guerra; Dolors Colomer

Clinical responses to bendamustine in chronic lymphocytic leukemia (CLL) are highly heterogeneous and no specific markers to predict sensitivity to this drug have been reported. In order to identify biomarkers of response, we analyzed the in vitro activity of bendamustine and the gene expression profile in primary CLL cells. We observed that mRNA expression of CD69 (CD69) and ITGAM (CD11b) constitute the most powerful predictor of response to bendamustine. When we interrogated the predictive value of the corresponding cell surface proteins, the expression of the activation marker CD69 was the most reliable predictor of sensitivity to bendamustine. Importantly, a multivariate analysis revealed that the predictive value of CD69 expression was independent from other clinico-biological CLL features. We also showed that when CLL cells were co-cultured with distinct subtypes of stromal cells, an upregulation of CD69 was accompanied by a reduced sensitivity to bendamustine. In agreement with this, tumor cells derived from lymphoid tumor niches harbored higher CD69 expression and were less sensitive to bendamustine than their peripheral blood counterparts. Furthermore, pretreatment of CD69 high CLL cases with the B-cell receptor (BCR) pathway inhibitors ibrutinib and idelalisib decreased CD69 levels and enhanced bendamustine cytotoxic effect. Collectively, our findings indicate that CD69 could be a predictor of bendamustine response in CLL patients and the combination of clinically-tested BCR signaling inhibitors with bendamustine may represent a promising strategy for bendamustine low responsive CLL cases.


British Journal of Haematology | 2017

Dual targeting of MCL1 and NOXA as effective strategy for treatment of mantle cell lymphoma.

Elisabeth Höring; Arnau Montraveta; Simon Heine; Markus Kleih; Lea Schaaf; Matthias Vöhringer; Anna Esteve-Arenys; Gaël Roué; Dolors Colomer; Elias Campo; German Ott; Walter E. Aulitzky; Heiko van der Kuip

Imbalances in the composition of BCL2 family proteins contribute to tumourigenesis and therapy resistance of mantle cell lymphoma (MCL), making these proteins attractive therapy targets. We studied the efficiency of dual targeting the NOXA/MCL1 axis by combining fatty acid synthase inhibitors (NOXA stabilization) with the CDK inhibitor Dinaciclib (MCL1 reduction). This combination synergistically induced apoptosis in cell lines and primary MCL cells and led to almost complete inhibition of tumour progression in a mouse model. Apoptosis was NOXA‐dependent and correlated with the NOXA/MCL1 ratio, highlighting the importance of the NOXA/MCL1 balance for effective cell death induction in MCL.


Molecular Cancer Therapeutics | 2011

Abstract A209: The nucleoside analogue acadesine exerts antitumoral activity and cooperates with anti-CD20 monoclonal antibodies in in vitro and in vivo models of mantle cell lymphoma.

Arnau Montraveta; Mercè de Frias; Clara Campàs; Elias Campo; Gaël Roué; Dolors Colomer

Mantle cell lymphoma (MCL) is a mature B-cell neoplasm characterized by the t(11;14)(q13:q32) that involves cyclin D1 overexpression and consequent cell cycle deregulation at the G1 phase. This entity is generally characterized by an aggressive course and a bad prognosis. Recently, a specific subtype of MCL has been described, showing best outcomes and that might be managed more conservatively than conventional MCL. These cases are characterized by non-nodal presentation, predominantly hypermutated IgVH, lack of genomic complexity, and absence of SOX11 expression. Acadesine is a nucleoside analogue initially developed as a cardioprotective agent, and which has shown a wide range of metabolic effects, including the activation of AMP-activated protein kinase (AMPK). Acadesine was shown to induce apoptosis in primary cells from several B lymphoid neoplasms and has been entered in a phase I/II clinical trial with relapsed/refractory chronic lymphocytic leukemia (CLL) patients. This clinical study has shown that acadesine plasmatic levels in the micro molar range are achievable and safe when CLL patients are treated with the drug. To evaluate the antitumoral properties of acadesine in MCL, we exposed a set of 11 MCL primary cultures and 9 MCL cell lines for up to 48h with increasing doses of the drug. Cytotoxicity and cytostatic effects were then assessed by flow cytometry detection of annexinV/propidium iodide labeling and MTT proliferation assay, respectively. In both MCL cell lines and MCL primary cultures, we observed a heterogeneous response to the drug, with no correlation to common genetic alterations such as deletion/mutation of P53, ATM or P16 genes. Rec-1, Jeko-1, UPN-1 and JVM-2 were the more sensitive cell lines, with a mean lethal dose 50 (LD50 of 1.57 mM at 24 h and 0.95 mM) at 48h, while 2 cell lines (HBL-2 and Granta-519) showed resistance to the compound (LD50 > 50 mM). Among MCL primary cultures, acadesine showed selective cytotoxic activity against malignant B cells while sparing accompanying T cells at pharmacologically achievable doses. Of note, those cases corresponding to the indolent MCL group showed increased sensitivity to the drug at 24h of treatment, when compared to conventional MCL cases (p=0.03). We observed that acadesine efficiently activates the intrinsic apoptotic pathway in MCL cells by reducing Mcl-1 levels, leading to conformational activation of Bax and Bak, mitochondrial depolarization, generation of reactive oxygen species and caspases processing. In drug combination assays, acadesine showed a synergistic effect when combined with the CD20 monoclonal antibody Rituximab. Finally, SCID mice were subcutaneously inoculated with 107 Jeko-1 cells. At day 12 post-inoculation, mice were randomized and administered for 18 days with either 400 mg/kg acadesine 5 days weekly, Rituximab 10mg/kg weekly, both drugs or vehicle. The combination was significantly more effective than Rituximab or Acadesine monotherapy (P Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A209.

Collaboration


Dive into the Arnau Montraveta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elias Campo

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Gaël Roué

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Laia Rosich

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge